You are on page 1of 11

ARTICLES

PUBLISHED: 28 NOVEMBER 2016 | VOLUME: 2 | ARTICLE NUMBER: 16226

A mouse model for MERS coronavirus-induced


acute respiratory distress syndrome
Adam S. Cockrell1*, Boyd L. Yount1, Trevor Scobey1, Kara Jensen1, Madeline Douglas1, Anne Beall1,
Xian-Chun Tang2,3, Wayne A. Marasco2,3, Mark T. Heise4,5*† and Ralph S. Baric1,4*†

Middle East respiratory syndrome coronavirus (MERS-CoV) is a novel virus that emerged in 2012, causing acute
respiratory distress syndrome (ARDS), severe pneumonia-like symptoms and multi-organ failure, with a case fatality rate
of ∼36%. Limited clinical studies indicate that humans infected with MERS-CoV exhibit pathology consistent with the late
stages of ARDS, which is reminiscent of the disease observed in patients infected with severe acute respiratory syndrome
coronavirus. Models of MERS-CoV-induced severe respiratory disease have been difficult to achieve, and small-animal
models traditionally used to investigate viral pathogenesis (mouse, hamster, guinea-pig and ferret) are naturally resistant
to MERS-CoV. Therefore, we used CRISPR–Cas9 gene editing to modify the mouse genome to encode two amino acids
(positions 288 and 330) that match the human sequence in the dipeptidyl peptidase 4 receptor, making mice susceptible
to MERS-CoV infection and replication. Serial MERS-CoV passage in these engineered mice was then used to generate a
mouse-adapted virus that replicated efficiently within the lungs and evoked symptoms indicative of severe ARDS,
including decreased survival, extreme weight loss, decreased pulmonary function, pulmonary haemorrhage and
pathological signs indicative of end-stage lung disease. Importantly, therapeutic countermeasures comprising MERS-CoV
neutralizing antibody treatment or a MERS-CoV spike protein vaccine protected the engineered mice against MERS-CoV-
induced ARDS.

T
he severity of respiratory illness caused by Middle East respir- MERS-CoV fails to replicate in traditional small-animal models
atory syndrome coronavirus (MERS-CoV), its pandemic (mouse, hamster, guinea-pig and ferret) due to the inability of the
potential through human-to-human respiratory transmission receptor-binding domain in the MERS-CoV spike protein to inter-
and a dearth of effective treatments necessitate the development act with the respective DPP4 receptor7–10. In addition to acting as
of new MERS-CoV therapies and vaccines. Effective vaccine and the MERS-CoV receptor, DPP4 regulates T-cell activation, cytokine
therapeutic development require preclinical animal models that function and trans-endothelial migration to sites of inflammation11.
resemble the pathogenesis of human MERS-CoV infection. Therefore, overexpression of DPP4 may result in immune dysregu-
Additionally, these models should: (1) include a measure of mor- lation. Effective models would therefore ideally promote functional
tality associated with severe respiratory disease; (2) not be con- MERS-CoV/DPP4 interactions, with minimal perturbations of
founded by neurological complications due to high viral loads in innate DPP4 expression, signalling activity or tissue distribution.
the brain; (3) exhibit sustained, high-level virus replication within Classical strategies to overcome receptor incompatibilities to gener-
the lungs of infected animals; (4) exhibit lung pathology associated ate susceptible mice have relied on generalized or tissue-specific
with human acute respiratory distress syndrome (ARDS); (5) main- transgenic overexpression approaches to drive expression of
tain innate expression of the MERS-CoV host receptor, dipeptidyl the human receptor (hDPP4) in the mouse12–15. Although
peptidase 4 (DPP4), to prevent perturbation of immunological MERS-CoV can elicit respiratory disease in hDPP4 overexpression
homeostasis; (6) be genetically tractable to study host genes that models, these models exhibit a fatal central nervous system (CNS)
regulate responses to MERS-CoV vaccines and therapeutics; and and systemic multi-organ disease12–14, probably due to non-specific
(7) exhibit reproducibility. overexpression of the receptor throughout the animal, which com-
Conventional non-human primate (NHP) models have been plicates the study of MERS-CoV-induced respiratory pathogenesis
established for MERS-CoV in both the rhesus macaque and in these models.
common marmoset1–4. NHPs are instrumental for the preclinical In this study, we used our knowledge of which determinants
development of therapeutics; however, these models are cost- allow mouse DPP4 to act as a functional MERS-CoV receptor7 by
prohibitive for initial screening of large numbers of vaccine and using CRISPR–Cas9 (clustered regularly interspaced short palindro-
therapeutic candidates, challenging to work with for routine mic repeats and CRISPR-associated gene 9) genome editing tech-
pathogenesis studies, limited in availability and typically require nology to insert codons that match the human sequence at
high virus challenge doses into multiple sites. Furthermore, two positions 288 and 330 in the mouse Dpp4 gene. This strategy
recent studies have contradicted the initial studies in NHPs, which resulted in a mouse that is permissive for MERS-CoV infection,
may complicate use of the rhesus macaque5 or the common while maximally preserving the species-specific interaction
marmoset6 model for routine vaccine or therapeutic testing. networks critical for DPP4 immune function. Generation of mice

1
Department of Epidemiology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA. 2 Department of Cancer Immunology and
AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA. 3 Department of Medicine, Harvard Medical School,
Boston, Massachusetts 02115, USA. 4 Departments of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North
Carolina 27599, USA. 5 Departments of Genetics, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA. † These authors jointly
supervised this work. * e-mail: adam_cockrell@unc.edu; mark_heisem@med.unc.edu; rbaric@email.unc.edu

NATURE MICROBIOLOGY 2, 16226 (2016) | DOI: 10.1038/nmicrobiol.2016.226 | www.nature.com/naturemicrobiology 1

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
ARTICLES NATURE MICROBIOLOGY

a b c 288/330+/+ 288/330+/− C57BL/6J WT

C57BL/6J WT
Exon 10 A288 Exon 11 T330

288/330+/+

288/330+/−
Exon 10 A288 Exon 11 T330
Lung
C57BL/6J WT

Exon 10 288L Exon 11 330R

Exon 10 A288 Exon 11 T330


288/330+/− Brain

Exon 10 288L Exon 11 330R

Exon 10 288L Exon 11 330R


288/330+/+
Kidney

d 1 × 109 288/330+/+ abc


288/330+/− def
1 × 108 C57BL/6J WT
a b e
Titre (p.f.u. ml−1 per g lung tissue)

d
1 × 107 c f
e 288/330+/+ 288/330+/− C57BL/6J WT
1 × 106

1 × 105 H&E

1 × 104

1 × 103

1 × 102 LOD IHC


1 × 101

1 × 100
EMC/2012 Camel MERS icMERS MERS-0

Figure 1 | A CRISPR–Cas9 genetically engineered mouse model for MERS-CoV replication. a, C57BL/6J mice were genetically engineered using
CRISPR–Cas9 genomic editing to encode 288L and 330R in mDPP4 on one chromosome (heterozygous, 288/330+/−) or on both chromosomes
(homozygous, 288/330+/+). b, Northern blot of mDPP4 mRNA expression. c, Immunohistochemistry (IHC) of mDPP4 protein in the lungs, brain and kidneys
of individual C57BL/6J wild-type (WT), 288/330+/− and 288/330+/+ mice. d, Viral titres for MERS-CoV at 3 days post-infection from C57BL/6J WT,
288/330+/− and 288/330+/+ (all n = 4) mice infected with 5 × 105 plaque-forming units (p.f.u.) of the indicated viruses. Bar graphs show means + s.d.
Student’s t-test was used to calculate P < 0.05 for comparisons of MERS-0 with each virus in 288/330+/+ mice (a–c on graph) and 288/330+/− mice
(d–f on graph). e, Pathology of 288/330+/+, 288/330+/− and C57BL/6J WT mice at day 3 after infection with MERS-0. Lung tissue sections were stained to
examine the pathology by haematoxylin and eosin staining (H&E), or were stained by IHC to detect nucleocapsid protein from MERS-0 infection. IHC and
H&E pathology images are representative of at least three samples. Scale bars (c,e), 1 mm.

carrying a chimaeric mouse DPP4 (mDPP4) molecule patterns in the lungs, kidneys and brains of 288/330+/+ and 288/
(A288L/T330R), combined with a mouse-adapted strain of MERS- 330+/− mice reflected those observed in C57BL/6J wild-type mice
CoV, allowed us to generate a mouse model that resembles severe (Fig. 1b,c; Supplementary Fig. 2). DPP4 is central to the
MERS-CoV-induced respiratory disease without bystander neuro- maintenance of glucose homeostasis in mammals16. Blood glucose
logical disease. In parallel, we demonstrated that this model system levels were within the normal range observed in C57BL/6J wild-
can be used for the development and testing of MERS-CoV vaccines type mice, supporting the hypothesis that biological mDPP4
and therapeutics. functions were not altered in the 288/330+/+ and 288/330+/− mice
(Supplementary Fig. 2). Moreover, basal CD4+ T-cell expression
Results of interleukin-2, tumour-necrosis factor-α, interferon-γ, CD69,
A CRISPR–Cas9-generated mouse model for MERS-CoV CD25 and mDPP4 (CD26) from the 288/330+/+ and 288/330+/−
infection. We have demonstrated previously that the introduction lines was comparable to the levels observed in C57BL/6J wild-type
of two amino acids that match the human sequence at positions mice (Supplementary Fig. 3). Notwithstanding functional T-cell
288 and 330 in the mDPP4 receptor can support MERS-CoV assessment, these results suggested that minimal alteration of the
docking, entry and replication in cell culture7. These determinants 288 and 330 alleles does not alter basal T-cell activation status.
are located within exons 10 and 11 of mDPP4 on chromosome 2 Overall expression levels, expression patterns, biological function
(Fig. 1a and Supplementary Fig. 1). Therefore, we used and the immunological profiles of mDPP4 were comparable to
CRISPR–Cas9 genome editing to introduce these determinants those of C57BL/6J wild-type mice following site-specific
(A288L and T330R) into the mDPP4 receptor (Fig. 1a and modification of the 288 and 330 alleles.
Supplementary Table 1). Two lines of C57BL/6J-derived mice The 288/330+/+ and 288/330+/− mice supported efficient infection
were generated that were either homozygous (288/330+/+) or and replication of the human MERS-CoV strain HCoV-EMC/2012,
heterozygous (288/330+/−) for the chimaeric mDPP4 alleles the camel MERS strain Dromedary/Al-Hasa-KFU-HKU13/2013
(Fig. 1a). The 288/330+/+ homozygous mice encoded the 288L and a recombinant virus derived from a molecular infectious
and 330R changes on both chromosomes, thereby expressing only clone (icMERS) in the lungs, but these virus strains could not repli-
mDPP4 with both changes (Fig. 1a). The 288/330+/− heterozygous cate in C57BL/6J wild-type mice that retained the original murine
mice encoded the 288L and 330R changes on one chromosome A288 and T330 alleles (Fig. 1d). In parallel, a MERS-CoV tissue-
and the C57BL/6J wild-type amino acids, A288 and T330, on the culture-adapted variant, derived by infection of NIH/3T3 cells ecto-
other chromosome, thereby expressing both mutated and wild- pically expressing the chimaeric mDPP4 (A288L/T330R) receptor,
type mDPP4 (Fig. 1a). The innate mDPP4 expression levels and was found to encode a three amino acid insertion (RMR) and a

2 NATURE MICROBIOLOGY 2, 16226 (2016) | DOI: 10.1038/nmicrobiol.2016.226 | www.nature.com/naturemicrobiology

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
NATURE MICROBIOLOGY ARTICLES
a 120 b 110

105
100
100

Weight relative to day 0 (%)


80 95
Survival (%)

90
60
85

40 80
288/330+/+, MERS-15
75
288/330+/−, MERS-15
20 288/330+/+, MERS-15
70 288/330+/+, MERS-0
288/330+/−, MERS-15 C57BL/6J WT, MERS-15
0 65
0 1 2 3 4 5 6 0 1 2 3 4 5 6
Day post-infection Day post-infection

c 1 × 109
Day 3
1 × 108 Day 6
d i ii
1 × 107
Titre (p.f.u. ml−1 per g lung tissue)

1 × 106

1 × 105

1 × 104
iii iv
1 × 103

1 × 102 LOD
< LOD
1 × 101

1 × 100
MERS-15 MERS-0 MERS-15 MERS-15
288/330+/+ 288/330+/+ 288/330+/− C57BL/6J WT

Figure 2 | Mouse-adapted MERS-CoV causes fatal disease in 288/330+/+ mice. Mice were inoculated intranasally with 5 × 106 p.f.u. a, Mortality of
288/330+/− (n = 10) and 288/330+/+ (n = 16) mice was monitored daily up to day 6 p.i. Data show the percentage of surviving mice. b, Mouse weights
were measured daily up to day 6 p.i. for 288/330+/+ mice infected with MERS-15 (n = 16) or MERS-0 (n = 10), 288/330+/− mice infected with MERS-15 (n = 10)
and C57BL/6J wild-type (WT) mice infected with MERS-15 (n = 7). Data are daily means of the percentage weight relative to day 0 ± s.d. c, Viral lung titres for
MERS-CoV were determined at day 3 p.i. (288/330+/− + MERS-15, n = 4; 288/330+/+ + MERS-15, n = 5; 288/330+/+ + MERS-0, n = 5; C57BL/6J WT + MERS-
15, n = 4) and day 6 p.i. (288/330+/− + MERS-15, n = 4; 288/330+/+ + MERS-15, n = 4; 288/330+/+ + MERS-0, n = 5; C57BL/6J WT + MERS-15, n = 3). The limit
of detection (LOD) is indicated. Bars are means + s.d. d, Immunohistochemistry of lung sections for anti-MERS nucleocapsid at 3 days p.i. Results show
288/330+/+ mice + MERS-15 (i), 288/330+/− mice + MERS-15 (ii), 288/330+/+ mice + MERS-0 (iii) and C57BL/6J WT mice + MERS-15 (iv). IHC images are
representative of at least three samples. Scale bar (d), 1 mm.

single amino acid change (S885L) in the S2 region of the spike gene. Mouse adaptation of MERS-CoV induces severe ARDS-like disease.
This recombinantly derived virus, MERS-0, encoding the RMR and The recombinantly derived MERS-0 virus was mouse adapted by
S885L S2 mutations, demonstrated significantly enhanced replica- serial passage for 15 rounds through the lungs in 288/330+/− mice
tion both in cell culture (Supplementary Fig. 4) and in the lungs at 3-day intervals, resulting in the MERS-15 strain. Infection of
of 288/330+/+ and 288/330+/− mice (Fig. 1d; P<0.05). Despite repli- 288/330+/+ mice via the intranasal route with MERS-15 resulted
cating to significantly higher virus titres in vivo than the other iso- in ∼70% mortality (genuine mortality, rather than mice meeting
lates, MERS-0 exhibited no evidence of severe clinical disease the typical 20% weight loss cut-off associated with humane
symptoms (Supplementary Fig. 4). Lung histology demonstrated euthanasia criteria), while 100% of infected 288/330+/− mice
that nucleocapsid antigen from MERS-0 (Fig. 1e), and from the survived (Fig. 2a). However, both lines exhibited 20–25% weight
other strains (not shown), was readily detected in the lungs of loss by day 6 p.i., in contrast to MERS-0-infected 288/330+/+ mice
infected mice by immunohistochemistry, but infected lungs exhib- or MERS-15-infected C57BL/6J wild-type mice, which exhibited
ited only moderate signs of respiratory pathology and inflammation. no weight loss (Fig. 2b). Significantly higher levels of MERS-15
These results demonstrated that we had developed a MERS-CoV replication were detectable in the lungs of both 288/330+/+ and
model that could support high levels of virus replication up to day 288/330+/− mice at days 3 and 6 p.i., while MERS-0 was mostly
3 post-infection (p.i.), but that further in vivo adaptation was cleared from the lungs by day 6 p.i. (Fig. 2c,d). Similar to MER-0,
required to achieve the respiratory symptoms characteristic of MERS-15 did not replicate in C57BL/6 wild-type mice.
MERS-CoV infection in humans. Importantly, the observed decreases in survival and weight loss

NATURE MICROBIOLOGY 2, 16226 (2016) | DOI: 10.1038/nmicrobiol.2016.226 | www.nature.com/naturemicrobiology 3

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
ARTICLES NATURE MICROBIOLOGY

a 50 b 0.8
MERS-15, 288/330+/+ MERS-15, 288/330+/+
MERS-15, 288/330+/− 0.7 MERS-15, 288/330+/−
MERS-0, 288/330+/+ MERS-0, 288/330+/+
** MERS-15, C57BL/6J WT 0.6 MERS-15, C57BL/6J WT
**
** 0.5

EF50 (ml s−1)


Penh(log10)

**
** ** **
5 ** 0.4
*
0.3

0.2 ** **
** * *
** ** 0.1 **
** **
0.5 0
0 1 2 3 4 5 6 0 1 2 3 4 5 6
Day post-infection Day post-infection

c i ii d i ii iii vii

iii iv iv v vi viii

Figure 3 | Lung function in MERS-15-infected mice. a,b, Respiratory function was monitored in live mice up to day 6 p.i. using whole-body plethysmography
to measure Penh (a) and EF50 (b) in 288/330+/+ mice infected with MERS-15 (n = 9) or MERS-0 (n = 3), 288/330+/− mice infected with MERS-15 (n = 4)
and C57BL/6J wild-type (WT) mice infected with MERS-15 (n = 3). Data are daily means ± s.d. Student’s t-test was used to compare 288/330+/+ mice
infected with MERS-15 and MERS-0 (*P < 0.05; **P < 0.01). c, Pathology of lungs from infected mice at day 3 p.i. demonstrating severe inflammation for
288/330+/+ (i) and 288/330+/− (ii) mice infected with MERS-15, and moderate inflammation for 288/330+/+ mice infected with MERS-0 (iii) and C57BL/6J
WT mice infected with MERS-15 (iv). d, Pathology at day 6 p.i. infected in mice. In 288/330+/+ mice infected with MERS-15, there was severe inflammation
and oedema in the large airways (i) and alveoli (ii), and hyaline membrane formation (iii). The 288/330+/− mice infected with MERS-15 exhibited severe
inflammation throughout the parenchyma (iv), hyaline membrane formation (v) and perivascular cuffing (vi). The 288/330+/+ mice infected with MERS-0 (vii)
and C57BL/6J WT mice infected with MERS-15 (viii) exhibited mild-to-moderate inflammation. Samples were stained with haematoxylin and eosin and are
representative of at least three samples. Scale bars (c,d), 1 mm.

induced by MERS-15 were not confounded by neurological mice infected with MERS-15 (Fig. 3a,b), demonstrating that
complications from brain infection, as plaque assays for MERS-15 elicited severe respiratory distress in mice carrying the
replication-competent virus and PCR with reverse transcription chimaeric DPP4 receptor. This was further supported by our obser-
(RT-PCR) at days 3 and 6 p.i. were negative (Supplementary vation of severe haemorrhage in the lungs of both 288/330+/+ and
Fig. 5). Moreover, quantitative RT-PCR on the same samples 288/330+/− mice infected with MERS-15 at days 3 and 6
demonstrated an increase of >106 detectable viral transcripts in (Supplementary Fig. 6), inflammatory infiltrates by day 3 (Fig. 3c)
infected lungs compared with similarly infected C57BL/6J mice, and respiratory pathology associated with severe acute respiratory
with no detectable viral transcripts in the brains of these mice distress, including hyaline membrane formation, intra-alveolar
(Supplementary Fig. 5c). oedema, perivascular cuffing and severe inflammation, at day
Although mortality and weight loss provide important measures 6 p.i. (Fig. 3d). Quantitative comparison of the lung pathology in
of MERS-CoV-induced disease, these parameters do not directly 288/330+/+ mice infected with MERS-15 and MERS-0 demonstrated
assess the impact of virus replication on respiratory function. that MERS-15 induced significant levels of pathology commensu-
Therefore, to directly assess the impact of MERS-15 infection on rate with ARDS by day 6 p.i. (Supplementary Fig. 7). Although
respiratory function in 288/330+/+ and 288/330+/− mice, we we did not conduct an exhaustive assessment of all extrapulmonary
measured respiratory function using unrestrained plethysmography, tissues in the 288/330+/+ mice, we could not detect virus replication
as demonstrated previously for respiratory pathogenesis in mouse in the brain, even at doses of 5 × 106 plaque-forming units (p.f.u.)
models of severe acute respiratory syndrome (SARS) and influ- up to day 6 when the humane euthanasia end points were
enza17. MERS-15 elicited severe lung disease as quantified by reached, and the pathology observed in our model was consistent
enhanced pause (Penh), a unitless measure that reflects airway with the severe respiratory pathology associated with fatal ARDS
obstruction/restriction due to debris in the airway, and by midtidal in the only published case study of a human MERS-CoV infection18.
expiratory flow (EF50), which represents the flow rate at which 50%
of the tidal volume has been expelled in a single breath17. MERS-15 Identification of MERS-CoV-adapted mutations associated with
infection led to significant increases in both Penh and EF50 in severe respiratory disease. We anticipated that the MERS-CoV
288/330+/+ and 288/330+/− mice up to day 6 p.i. compared with genome would acquire mutations due to immunological pressure
288/330+/+ mice infected with MERS-0 and C57BL/6J wild-type and/or enhanced virus fitness during mouse adaptation. Two viral

4 NATURE MICROBIOLOGY 2, 16226 (2016) | DOI: 10.1038/nmicrobiol.2016.226 | www.nature.com/naturemicrobiology

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
NATURE MICROBIOLOGY ARTICLES
a 120 b 110

105
100

Weight relative to day 0 (%)


100

80 95
Survival (%)

90
60
85

40 80
MERS-15 C1, 288/330+/+
MERS-15 C1, 288/330+/+ 75 MERS-15 C2, 288/330+/+
20 MERS-0, 288/330+/+
MERS-15 C2, 288/330+/+ 70 MERS-15 C2, C57BL/6J WT

0 65
0 1 2 3 4 5 6 0 1 2 3 4 5 6
Day post-infection Day post-infection
c 1 × 109
Day 3
1 × 108 Day 6
Titre (p.f.u. ml−1 per g lung tissue)

1 × 107 d i ii iii

1 × 106

1 × 105

1 × 104
iv v vi
1 × 103

1 × 102 LOD
< LOD
1 × 101

1 × 100
MERS-15 C1 MERS-15 C2 MERS-0 MERS-15 C2
288/330+/+ 288/330+/+ 288/330+/+ C57BL/6J WT

Figure 4 | Clonal isolates of mouse-adapted MERS-CoV exhibit severe respiratory disease. Mice were inoculated intranasally with 5 × 106 p.f.u. a, The
mortality of 288/330+/+ mice infected with MERS-15 C1 (n = 7) or MERS-15 C2 (n = 11) was monitored daily up to day 6 p.i. Data reflect the percentage of
surviving mice. b, Mouse weights were monitored daily for 288/330+/+ mice infected with MERS-15 C1 (n = 7), MERS-15 C2 (n = 11) or MERS-0 (n = 6), and
C57BL/6J wild-type (WT) mice infected with MERS-15 C2 (n = 6). Data are daily means ± s.d. c, Viral lung titres were determined at day 3 p.i. (288/330+/+ +
MERS-15 C1, n = 4; 288/330+/+ + MERS-15 C2, n = 3; 288/330+/+ + MERS-0, n = 3; C57BL/6J WT + MERS-15 C2, n = 3) and day 6 p.i. (288/330+/+ +
MERS-15 C1, n = 4; 288/330+/+ + MERS-15 C2, n = 6; 288/330+/+ + MERS-0, n = 3; C57BL/6J WT + MERS-15 C2, n = 3). The limit of detection (LOD) is
indicated. Bars are means + s.d. d, IHC of lung sections at 3 days p.i. from 288/330+/+ mice infected with MERS-15 C1 (i) or MERS-15 C2 (ii) and stained for
nucleopcapsid. The pathology of the lungs from 288/330+/+ mice infected with MERS-15 C2 was assessed by haematoxylin and eosin staining (H&E) at day
6 p.i. and demonstrated severe inflammation (iii), oedema (iv), hyaline membrane formation (v) and perivascular cuffing (vi). All H&E images are
representative of at least three samples. Scale bar (d), 1 mm.

clones, MERS-15 clone 1 (MERS-15 C1) and MERS-15 clone 2 orf4b into orf5 (Supplementary Fig. 9). Moreover, 5′ rapid
(MERS-15 C2), were isolated by plaque purification from the amplification of cDNA ends revealed that the nucleotide at
MERS-15 heterogeneous virus population. MERS-15 C2 infection position 2 of the 5′ untranslated region was deleted in both clones
resulted in increased mortality of the mice (Fig. 4a) and (Supplementary Fig. 9). Generation of an infectious clone
significantly increased haemorrhage up to day 6 p.i. compared harbouring all of the MERS-15 C2 mutations (icMERSma1)
with MERS-15 C1 (Supplementary Fig. 8), whereas both clonal demonstrated that the disease could be reproduced with an
isolates caused 25–30% weight loss (Fig. 4b) and high levels of infectious dose of 5 × 106 p.f.u. (Supplementary Fig. 10). Decreasing
virus replication in the mice up to day 6 p.i., when humane the dose by 10-fold to 5 × 105 p.f.u. resulted in weight loss that
euthanasia end points were reached (Fig. 4c). The lung pathology paralleled that observed with 5 × 106 p.f.u.; however, the 5 × 105 p.f.u.
elicited by MERS-15 C2 resembled that obtained with the primary dose exhibited a slight decrease in mortality up to day 7 p.i.
MERS-15 virus (Fig. 4d), demonstrating similar pathologies (Supplementary Fig. 10). Although additional studies will be
including oedema, hyaline membrane formation and perivascular needed, comparative genomic analysis of the two clones, C1 and
cuffing. Sequencing of the entire MERS-15 C2 genome revealed a C2, indicated that nsP2 and Orf5 may have significant roles in
set of unique missense mutations, acquired during in vivo determining disease outcome. Therefore, the MERS-15 C2 derived
passaging, in the genes encoding the non-structural proteins, virus, combined with the 288/330+/+ mouse line, represent useful
nsP2, nsP6 and nsP8, and a large deletion in orf4b that may be tools for studying MERS-CoV pathogenesis or assessing
responsible for the enhanced disease observed with MERS-15 therapeutic countermeasures against MERS-CoV-induced ARDS.
(Supplementary Fig. 9). Sequencing of MERS-15 C1 revealed
some differences that may influence the capacity of the virus to Human monoclonal antibody 3B11 protects from MERS-CoV-
elicit the increased mortality observed with MERS-15 C2, namely elicited severe respiratory disease. Human monoclonal antibodies
mutations in nsP2 and an expanded deletion that extended from provide a robust strategy for the treatment of newly emerged viruses

NATURE MICROBIOLOGY 2, 16226 (2016) | DOI: 10.1038/nmicrobiol.2016.226 | www.nature.com/naturemicrobiology 5

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
ARTICLES NATURE MICROBIOLOGY

a 120 b
105

100 100

Weight relative to day 0 (%)


95
80
Survival (%)

90
60
85

40 80

75
20 3B11 3B11
70
Isotype control Isotype control
0 65
0 1 2 3 4 5 6 0 1 2 3 4 5 6

Day post-infection Day post-infection

c 1 × 109 d
Day 3 3B11
16
1 × 108 Day 6 Isotype control
14 **
Titre (p.f.u. ml−1 per g lung tissue)

1 × 107
12
1 × 106

10
1 × 105 Penh

1 × 104 8

1 × 103 6
*

1 × 102 LOD 4
< LOD
1 × 101 2
** *
1 × 100 0
Isotype control 3B11 0 3 6
Day post-infection

Figure 5 | Human monoclonal antibody 3B11 protects mice from severe respiratory disease. The 288/330+/+ mice were intraperitoneally administered
250 µg of 3B11 human monoclonal antibody or isotype control antibody 12 h before challenge with 5 × 106 p.f.u. MERS-15 C2. a, b, Mortality (a) and mouse
weight (b) were monitored daily for mice receiving 3B11 (n = 12) or an isotype control (n = 12) up to day 6 p.i. Data show the percentage of surviving
mice (a) or daily mean weight ± s.d. (b). c, Viral lung titres were determined at day 3 p.i. (n = 6 for both 3B11- and isotype control-treated mice) and day 6 p.i.
(n = 6 for 3B11-treated mice; n = 3 for isotype control-treated mice). The limit of detection (LOD) is indicated. Bars are means + s.d. d, Lung function was
assessed by Penh at 0, 3 and 6 days p.i. for mice receiving 3B11 (n = 6 at days 0, 3 and 6) or isotype control (n = 6 at days 0 and 3; n = 3 at day 6). Data are
means + s.d. Student’s t-test was used to compare mice receiving 3B11 with those receiving the isotype control at day 3 and day 6 p.i. (*P < 0.05; **P < 0.01).

in humans. 3B11 is a human monoclonal antibody that targets the particles (VRPs) expressing MERS-CoV spike protein (spike–VRP)
receptor-binding domain of the MERS-CoV spike protein19, and is or mock vaccinated with VRPs expressing green fluorescent protein
effective in NHPs5. As the MERS-15 C2 adapted virus acquired no (GFP–VRP), boosted at 4 weeks postprime and challenged with
receptor-binding domain mutations (Supplementary Fig. 9), we MERS-15 C2 at 4 weeks postboost. All mice receiving spike–VRP
reasoned that 3B11 should protect 288/330+/+ mice from MER-15 survived and exhibited no weight loss following challenge compared
C2 challenge. Pretreating mice for 12 h with 3B11 provided 100% with GFP–VRP mock-vaccinated animals (Fig. 6a,b). Spike–VRP
protection against MERS-15 C2 challenge (Fig. 5a,b). Moreover, vaccination significantly reduced MERS-15 C2 replication in the
3B11 treatment reduced viral loads in the lungs of infected mice to lungs of infected mice as shown by both plaque titre (Fig. 6c) and
undetectable levels (Fig. 5c and Supplementary Fig. 11) and viral antigen staining (Supplementary Fig. 14), while also protecting
protected from loss of respiratory function (Fig. 5d) (Supplementary against severe respiratory disease, as assessed by measurement of
Fig. 11), pulmonary haemorrhage (Supplementary Fig. 11) and Penh (Fig. 6d) and EF50 (Supplementary Fig. 13), lung haemorrhage
severe pathological changes (Supplementary Fig. 12). In contrast, (Supplementary Fig. 13) and pathological indications of severe acute
pretreatment with isotype control antibody provided no protective respiratory disease (Supplementary Fig. 14). Neutralization of
effect. Therefore, these data convincingly demonstrated that our MERS-15 C2 with prechallenge serum from spike–VRP-vaccinated
preclinical mouse model of severe respiratory disease and mortality mice validated the presence of high-titre neutralizing antibodies in
can serve as a platform for assessing MERS-CoV therapeutics. the serum of vaccinated 288/330+/+ mice (Supplementary Fig. 13).
Our data demonstrated that the spike–VRP vaccine provoked an
Spike protein vaccines derived from Venezuelan equine adaptive immune response capable of protecting mice from a lethal
encephalitis virus replicon particles protect from lethal infection. challenge with MERS-CoV, thereby extending the utility of our
To examine vaccine efficacy in the 288/330+/+ MERS-15 C2 model, preclinical mouse model of severe respiratory disease to include
mice were vaccinated with Venezuelan equine encephalitis replicon vaccine evaluation.

6 NATURE MICROBIOLOGY 2, 16226 (2016) | DOI: 10.1038/nmicrobiol.2016.226 | www.nature.com/naturemicrobiology

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
NATURE MICROBIOLOGY ARTICLES
a 120 b
110

100 105

Weight relative to day 0 (%)


100
80
95
Survival (%)

60 90

85
40 80

75
20
Mock vaccinated (GFP−VRP) Mock vaccinated (GFP−VRP)
70
Vaccinated (spike−VRP) Vaccinated (spike−VRP)
0 65
0 1 2 3 4 5 6 0 1 2 3 4 5 6
Day post-infection Day post-infection

c 1 × 109 d
Day 3 12 Mock vaccinated (GFP−VRP)
1 × 108 Day 6 ** Vaccinated (spike−VRP)
Titre (p.f.u. ml−1 per g lung tissue)

10
1 × 107

1 × 106
8 *
1× 105
Penh 6
1 × 104

1 × 103 4 **

1 × 102 LOD
< LOD 2
1 × 101 *

1 × 100 0
GFP−VRP Spike−VRP 0 3 6
Day post-infection

Figure 6 | Vaccination of 288/330+/+ mice with a VRP delivering MERS-CoV spike protein protects mice from challenge with MERS-CoV. The vaccination
protocol is described in Methods. a,b, After a 5 × 106 p.f.u. challenge, mortality (a) and mouse weight (b) were monitored daily for mice receiving GFP–VRP
(n = 19) or spike–VRP (n = 19) vaccine up to day 6 p.i. Data reflect the percentage survival (a) or daily mean weight ± s.d. (b). c, Viral lung titres were
determined on day 3 p.i. (n = 7 for spike–VRP and GFP–VRP) and day 6 p.i. (n = 12 for spike–VRP; n = 6 for GFP–VRP). The limit of detection (LOD) is
indicated. Bars are means + s.d. d, Lung function was assessed by Penh at 0, 3 and 6 days p.i. for mice receiving GFP–VRP (n = 12 at days 0 and 3 p.i.; n = 6
at day 6 p.i.) or spike–VRP (n = 12 at all days p.i.). Data represent means + s.d. Student’s t-test was used to compare mice receiving GFP–VRP with those
receiving spike–VRP at days 3 and 6 p.i. (*P < 0.05; **P < 0.01).

Discussion reported histopathology that included diffuse alveolar damage with


The MERS-CoV preclinical mouse model described here demon- denuding of bronchiolar epithelium, hyaline membrane formation,
strates for the first time that the CRISPR–Cas9 system can be used type II pneumocyte hyperplasia and oedema18. Furthermore,
to genetically edit a non-permissive host receptor to generate a sus- MERS-CoV antigen staining localized the virus to pneumocytes
ceptible model for an emerging infectious pathogen. The 288/330+/+ and syncytial cells18. Infection of type I and II pneumocytes can
MERS-CoV mouse model resembles the severe, and often fatal, res- lead to cell death, as observed in autopsies of patients who have
piratory distress syndrome observed in humans, and can be pre- died from severe respiratory infections with influenza virus and
vented through treatment with the 3B11 neutralizing monoclonal SARS-CoV21,22. Moreover, pneumocyte cell death has been proposed
antibody19 or a VRP-based vaccine directed against the to cause decreased respiratory function, as measured by whole-body
MERS-CoV spike protein15,20. Coupled with an inability to elicit plethysmography in a mouse model of influenza23. Commensurate
escape mutants, 3B11 has also been tested in NHPs5, making it an with these previous studies, our MERS-CoV mouse model demon-
excellent candidate for downstream human studies. However, exist- strated widespread infection of pneumocytes and pathology consist-
ing NHP models rely on quantitative RT-PCR, rather than measures ent with diffuse alveolar damage and severe respiratory disease. This
of infectious virus, to quantify viral loads, and these models do not was corroborated by decreased pulmonary function in the
reproducibly result in the severe respiratory disease or mortality MERS-CoV model, as measured by plethysmography, which may
observed in human MERS patients1–4. Therefore, the results from be associated with widespread infection and possibly the death of
our model complement the NHP 3B11 studies by directly demon- pneumocytes and airway epithelial cells. Therefore, this model
strating that 3B11 reduces levels of infectious MERS-CoV in the system provides the field with the opportunity to investigate the
lungs, while preventing severe virus-induced respiratory pathology, mechanisms that lead to MERS-CoV-induced pathology and
loss of respiratory function and mortality. severe respiratory disease, in the absence of any CNS complications.
In fatal cases of human MERS-CoV infections, individuals Other MERS-CoV mouse models have used the more traditional
exhibit severe respiratory distress requiring mechanical ventilation, method of expressing the full-length human DPP4 receptor to facili-
and the only published human autopsy from a MERS-CoV fatality tate MERS-CoV infection12–15,24. These models exhibited infection/

NATURE MICROBIOLOGY 2, 16226 (2016) | DOI: 10.1038/nmicrobiol.2016.226 | www.nature.com/naturemicrobiology 7

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
ARTICLES NATURE MICROBIOLOGY

replication in the lungs following intranasal administration at low models to evaluate therapeutic countermeasures, thereby necessitat-
viral doses (102–105 p.f.u.), which in some cases resulted in pathol- ing virus adaptation to host immunity to achieve effective models. It
ogy indicative of pneumonia-like disease12–14. One limitation of the is critical that the GOF regulatory structure does not impede the
288/330+/+ MERS-CoV model described here was the use of high development of robust animal models of human disease, which
viral loads to achieve severe, and often fatal, respiratory disease. are essential for protecting the public health.
Further adaptation may allow the use of lower infectious doses
that would produce a model of mild disease with subsequent recov- Methods
ery at later time points, as has been described with SARS-CoV25. In Viruses, cells and plaque assays. All virus stocks were prepared on Vero CCL81
this context, it is interesting that the deletion of MERS-CoV ORF4b, cells (ATCC). CCL81 cells were maintained routinely in Dulbecco’s modified Eagle’s
medium (Gibco) supplemented with 10% fetal bovine serum (FBS; Sigma) and
a phosphodiesterase and antagonist of RNase L activity in human 1× antibiotic/antimycotic (Gibco). All viruses were harvested in Opti-MEM medium
cells, appeared less critical for eliciting severe disease in rodents26, (Gibco) supplemented with 3% FBS, 1× antibiotic/antimycotic, 1× non-essential
suggesting possible species-specific modes of action in vivo. amino acids (Gibco) and 1× 1 mM sodium pyruvate (Gibco). The wild-type HCoV-
Deletions in some SARS-CoV interferon antagonist genes and EMC/2012 strain of MERS-CoV was used at passage 10 (originally provided by Bart
Haagmans (Erasmus Medical Center Rotterdam, The Netherlands) at passage 8),
accessory open reading frames have also yielded subtle changes in
icMERS was generated previously by Scobey et al. 33, MERS camel strain Dromedary/
overall virulence in vivo 27,28. However, it is important to point out Al-Hasa-KFU-HKU13/2013 was used at passage 5 and was provided at passage 4 by
that a balance must be achieved between the virulence of mouse- Malik Peiris (University of Hong Kong, China) and MERS-0 was generated in the
adapted virus, which enhances the model’s capacity to replicate laboratory of R.S.B. as described below. Virus titres were determined by plaque
human disease phenotypes, and the number of mutations required assays on Vero CCL81 cells33. All viruses were maintained under Biosafety Level
(BSL) 3 conditions with redundant fans and personnel powered air-purifying
to significantly reduce the dose lethal to 50% of animals tested. respirators, scrubs, Tyvek suits, Tyvek aprons and double layers of gloves.
Conventional models using constitutive overexpression of the An icMERS virus tagged with tomato red fluorescent protein33 was passaged for
hDPP4 MERS-CoV receptor have demonstrated widespread infec- ten rounds on NIH/3T3 cells (ATCC) that were generated to stably overexpress the
tion of extrapulmonary tissues including brain, kidney, liver, mDPP4 receptor containing A288L and T330R. The mDPP4 A288L/T330R
spleen and heart12–14, and two of these studies indicated that the expression cassette used to generate the cell line has been described previously7.
Sequencing of the passaged virus identified an insertion of three amino acids (RMR)
mice exhibited multi-organ failure13,14. Furthermore, high viral after amino acid 884 in the spike protein and an S885L change. These changes were
loads were detected in the brains of mice in the transgenic hDPP4 subcloned back into the MERS-CoV infectious clone by overlap-PCR of the F
overexpression models12–14. Importantly, Li et al. 13 concluded that fragment with the following primers: 5′-GGTTTCCAGAAGTGTGAGCAATTA
“mortality correlated with brain infection, suggesting that infection CTGCGCG-3′, 5′-GCAGGCCTCTGCAGTCGACGGGCCCGGGATCCAA
of this organ was most important for the high mortality observed in TGCC-3′, 5′-CCTGTTTCTATATCTACTGGCAGTCG TAGAATGCGGCTTGCA
CGTAGTGCTATTGAGGATTTGC-3′ and 5′-GCAAATCCTCA ATAGCACTA
K18-hDPP4 mice”. While these lethal models have value for vaccine CGTGCAAGCCGCATTCTACGACTGCCAGTAGATATAGAAACAGG-3′. The F
and immunotherapeutic testing29, small-molecule inhibitors that are fragment is one part of a seven-plasmid system (A, B, C, D1, D2, E and F) that
effective in the lung may be limited in their efficacy due to an permits partitioning of the entire genome to generate infectious MERS-CoV, as
inability to cross the blood–brain barrier30. Similar neurological described previously33. The PCR product encoding the insertion was subcloned back
complications have been observed in model systems for SARS that into the F fragment using the restriction enzymes MscI and BamHI and validated by
sequencing. Recombinant MERS-0 virus was used for the in vivo passage
used overexpression, or tissue-specific constitutive promoters, to experiments, which were GOF studies reviewed and approved by the National
express human angiotensin 1-converting enzyme 2 (ACE2) in Institutes of Health (NIH). Vero CCL81 and NIH/3T3 cells were originally received
mice31. While additional human pathology studies are needed to from the ATCC, which indicates that cell lines are authentic and confirms that cell
determine the extent of extrapulmonary sites of MERS-CoV replica- lines are mycoplasma free. None of the working cell line stocks was authenticated or
tion and their impact on MERS-CoV disease, it is clear that respir- tested for mycoplasma recently, although the original seed stocks used to create the
working stocks are free from contamination. Additionally, both cell lines have been
atory replication and pathology is an important aspect of human authenticated by morphological and cytopathological evaluation. Furthermore, Vero
MERS-CoV disease. Therefore, an important attribute of the CCL81 cells were confirmed for DPP4 overexpression by the capacity to be infected
288/330+/+ model is that the lack of detectable virus replication in and to replicate MERS-CoV.
the CNS means this model can be used to study MERS-CoV-
induced pulmonary disease without the confounding effects of Generation of mice with mDPP4 modified at positions 288 (exon 10) and
330 (exon 11). The alleles encoding amino acids 288 and 330 are shown in
death due to CNS infection.
Supplementary Fig. 1. Genomic engineering of these alleles with the CRISPR–Cas9
Recently, a debate has emerged around the safety of performing genome editing system was performed at the University of North Carolina at Chapel
gain-of-function (GOF) studies with highly pathogenic viruses Hill (UNC-CH) Animal Models Core Facility. The messenger RNA (mRNA)
(such as MERS-CoV, SARS-CoV, influenza virus H5N1) (http:// encoding Cas9 endonuclease and guide RNAs (gRNAs) were based on the system
www.gryphonscientific.com/gain-of-function/). As demonstrated established by Mali et al.34 and prepared as described below. The gRNAs
(Supplementary Table 1) were generated by an in vitro transcription reaction using a
here, GOF studies were absolutely necessary to develop a mouse T7 High Yield RNA Synthesis kit (NEB), where 1 µg DraI-linearized template DNA
model that reflects the ARDS pathology observed previously in was used in a 20 µl reaction following the kit guidelines for short RNA transcripts.
humans infected with respiratory pathogens. Importantly, the The reaction was incubated at 37 °C overnight, followed by DNase I (RNase-free)
GOF studies performed here yielded MERS-CoV strains that digestion for 15 min at 37 °C. The gRNAs were then purified using an RNeasy
reflect the complexity of clinical isolates identified recently in column (Qiagen) following the guidelines for short RNA purification. Capped and
polyadenylated Cas9 mRNA was prepared by an in vitro transcription reaction using
humans, where deletions were identified in ORF3 and ORF4A32. an mMESSAGE mMACHINE T7 ULTRA kit (Life Technologies). Capped mRNA
Moreover, these GOF studies have allowed us to identify mutations was generated with 1 µg hCas9-T7 linearized plasmid DNA in a 20 µl reaction
in MERS-CoV proteins that may influence how MERS-CoV inter- containing 1× NTP/ARCA Solution, 1× T7 reaction buffer and 2 µl T7 enzyme. The
acts with, and possibly circumvents, host immune responses. reaction was incubated at 37 °C for 1 h, followed by addition of 1 µl TURBO DNase
Nevertheless, future studies to evaluate host factors that contribute and digestion at 37 °C for 15 min. To add a poly(A) tail to the capped mRNA, the
20 µl reaction mix from step 1 was mixed with nuclease-free water (36 µl), 5× E-PAP
to MERS-CoV disease will be constrained in this model, as well as buffer (20 µl), 25 mM MnCl2 (10 µl), ATP solution (10 µl) and E-PAP (4 µl) to a
in hDPP4 expression models, by the fact that the mice must be back- final reaction volume of 100 µl. The reaction was incubated at 37 °C for 30–45 min.
crossed to mouse lines harbouring modified endogenous genes, The capped and polyadenylated RNA was purified by lithium chloride precipitation
such as knock-out mice. This limitation may be overcome and resuspended in microinjection buffer (5 mM Tris/HCl buffer, 0.1 mM EDTA,
through additional GOF studies that facilitate MERS-CoV adap- pH 7.5).
Fertilized zygotes were collected from C57BL/6J females that had been
tation to the innate mDPP4 receptor molecule. The continued superovulated and mated to C57BL/6J males. Pronuclear microinjection was
threats from novel emerging pathogens, such as Zika virus, will performed with 50 ng µl−1 Cas9 mRNA, 50 ng µl−1 gRNA Dpp4-g59B, 25 ng µl−1
demand the rapid development of physiologically relevant animal gRNA Dpp4-g88B and 50 ng µl−1 each of Dpp4-A288L-B and Dpp4-T330R-B

8 NATURE MICROBIOLOGY 2, 16226 (2016) | DOI: 10.1038/nmicrobiol.2016.226 | www.nature.com/naturemicrobiology

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
NATURE MICROBIOLOGY ARTICLES
donor oligonucleotides (Supplementary Table 1). Injected embryos were implanted day 0 to establish a baseline and again at days 3 and 6 p.i. The isolation and
into pseudopregnant recipients, and the resulting pups were screened for alleles production of the 3B11 and F10 antibodies has been described previously19.
encoding changes (Supplementary Fig. 1) at positions 288 and 330. Mutations at
the 288 position were detected by amplifying biopsy DNA samples with the Spike–VRP vaccine study and virus neutralization assay. The MERS-CoV gene
following primers: Dpp4-E10ScF1: 5′-GATTCTGAGCAAGCAAACACGC-3′, encoding the spike protein and GFP gene were packaged into VRPs generated with
and Dpp4-E10ScR1: 5′-CCACAAGGTATCCCACAGAGACG-3′. The helper constructs from the V3526 attenuated strain of Venezuelan equine
752 bp PCR product was sequenced with primer Dpp4-E10-SqR1: encephalitis virus, under BSL2 conditions, as described previously20. Mice were
5′-CAAGAACCACACCAATGGAAAGTC-3′. Mutations at the 330 position were administered a primary vaccination of 10 µl by footpad injection of spike–VRP
detected by amplifying biopsy DNA samples with the following primers: (1 × 105 p.f.u.) or control GFP–VRP (1 × 105 p.f.u.). After 28 days, the mice were
Dpp4-E11ScF1: 5′-AAGTGCTGGGATTATAGGTGGTCAC-3′, and Dpp4-E11ScR1: boosted with the same dose of their respective VRP strain. At 28 days postboost, all
5′-GTGTTTACATTCTAAGTTGGGTTTCTGC-3′. The 767 bp PCR product was vaccinated mice were challenged with 5 × 106 p.f.u. MERS-15 C2. Mice were
sequenced with primer Dpp4-E11-SqF1: 5′- GCATGTTATCCACTGTGCCATCTC-3′. monitored daily for weight and survival, and killed at days 3 and 6 p.i. to collect
Five of 66 live animals produced showed evidence of both the 288 and 330 modifications. lungs for histology, assessment of viral titre by plaque assay and evaluation of lung
Founders with both expected modifications were backcrossed to C57BL/6J mice to haemorrhaging. Respiratory function was measured at day 0 to establish a baseline
identify animals with the 288 and 330 modifications in cis. F1 animals with both the 288 and then again at days 3 and 6 p.i.
and 330 modifications were then intercrossed to produce homozygous breeder pairs for The presence of MERS-15 C2-specific serum antibodies was assessed by a plaque
colony enrichment and downstream studies. reduction neutralization titre assay. Prechallenge serum samples were collected at
25 days postboost with the spike–VRP vaccine or the GFP–VRP control. Virus
Mouse infections. Genetically engineered mice with a modified mDPP4 receptor neutralization assays were performed as described previously35. The percentage of
were housed and bred in accordance with guidelines established by the Department plaque reduction was calculated as: 1 − (number of plaques with spike–VRP or
of Laboratory Animal Medicine at UNC-CH. As the 288/330+/+ and 288/330+/− mice GFP–VRP serum/number of plaques with serum from naïve 288/330+/+ mice) × 100.
are novel mouse lines developed in the laboratory of R.S.B., experiments utilized
available male and female mice that ranged from 12 to 20 weeks of age. Based on Respiratory function. Respiratory function was measured for individual mice, at the
availability at the time of each experiment, experimental and control animals were indicated time points, as demonstrated previously for SARS-CoV and influenza A
age- and sex-matched. No blinding was used in any animal experiments, and virus17. Briefly, individual mice were acclimated for 30 min in individual
animals were not randomized. Sample sizes were determined from preliminary data plethysmography chambers (Buxco Systems) and each breath was then quantified
that would yield statistically significant differences. Mouse studies were executed over a 5 min period. Mice were routinely randomized into different chambers to
under animal BSL3 conditions as described previously35. Before viral infection, mice avoid measurement biases that could result between chambers. Data for Penh and
were anesthetized by administering 50 µl ketamine/xylazine mixture EF50 were analysed as described previously by our group17.
intraperitoneally and then infected intranasally with 50 µl virus solution containing
5 × 106 p.f.u. Incomplete infections due to bubbling of inoculum from the nasal Histology. Lung, brain and kidney samples were placed in 10% phosphate-buffered
cavity, the inability to inhale the entire dose or inoculum going into the mouth were formalin for >7 days at 4 °C for fixation. Fixed tissue samples were then removed
noted, and these mice were considered failures and were excluded, as described from the BSL3, placed into cassettes for embedding in paraffin and submitted to the
previously35. Following sedation and infection, mice were monitored daily for weight Lineberger Comprehensive Cancer Center Animal Histopathology Core for
loss and survival, as well as for signs that the animals were moribund (including processing, sectioning and staining. Tissue sections (5 µm) were stained with
laboured breathing, lack of movement and lack of grooming). Mice that reached 20% haematoxylin and eosin, and MERS-CoV nucleocapsid antigen was detected using
weight loss were placed under exception and monitored at least twice daily. Mice that mouse anti-MERS nucleocapsid serum at a 1:250 dilution. The nucleocapsid
approached 30% weight loss were euthanized immediately. Mice deemed moribund antiserum was generated in the laboratory of R.S.B. using MERS-CoV nucleocapsid–
were euthanized at the discretion of the researcher. Mice were euthanized with an VRP particles in BALB/c mice as described previously20. Antigen was visualized
isoflurane overdose followed by a secondary thoracotomy, at various time points, to using 3,3′-diaminobenzidine staining. An Olympus DP71 camera attached to an
collect lung tissues. In the absence of a thoracotomy, cervical dislocation was used as Olympus BX41 microscope was used to capture images with ×10 and ×40 objectives.
a secondary euthanasia method. All are approved methods of the Institutional Histopathology was scored, blinded to infection and animal status, for airway
Animal Care and Use Committee (IACUC) at the UNC-CH. disease, vascular disease, parenchymal pneumonia, diffuse alveolar damage,
eosinophils and immunohistochemistry on a scale of 0–3 (0, none; 1, mild; 2,
Ethics statement. Mouse studies were carried out in accordance with the moderate; 3, severe).
recommendations for the care and use of animals by the Office of Laboratory
Animal Welfare at NIH. IACUC at UNC-CH approved the animal studies Flow cytometry analysis. Whole peripheral blood from each mouse strain was
performed here (protocol, IACUC 13-272), using a weight loss cut-off point of collected in EDTA and peripheral blood mononuclear cells were purified using
∼30% for humane euthanasia. Synthetically reconstructed MERS-CoVs were density-gradient centrifugation over Ficoll-Paque PLUS (GE Healthcare Life
approved by the UNC-CH Institutional Biosafety Committee, which also considered Sciences). Mouse lung tissues were first dissociated mechanically using scalpel blades
GOF research concerns before execution of these experiments. and then digested further using type A collagenase (Worthington Biochemical) in
medium prepared with DNase I (1 mg ml−1) for 1 h in a shaking incubator at 37 °C.
Analysis of serum glucose levels. All blood glucose measurements were taken Digested lung suspensions were filtered, centrifuged and treated with ACK lysis
following a 6 h fast. Blood glucose was measured by tail clip sampling using an buffer (Gibco). Single-cell suspensions from blood and lung tissue were resuspended
AlphaTRAK 2 glucometer (Abbott Laboratories)36. This system is designed for use at 1 × 107 cells ml−1 in RPMI 1640 medium supplemented with 10% heat-inactivated
in laboratory mice, with a normal range of 111–205 mg dl−1 (6.1–11.38 mmol l−1) FBS and antibiotic/antimycotic cocktail (all from Gibco), and were used
blood glucose and a detection limit of 20–750 mg dl−1 (1.1–41.62 mmol l−1) immediately for flow cytometry. Single-cell suspensions were plated in 200 µl per
blood glucose. well on 96-well, round-bottomed plates and stained for flow cytometry using an
Intracellular Fixation and Permeabilization Buffer Set (eBioscience) according to the
Adaptation of MERS-0 in humanized mice. The recombinant virus MERS-0 was manufacturer’s protocol and using antibodies specific for CD3 (clone 145-2C11; BD
passed through the lungs of 288/330+/− mice every 3 days for 15 passages to obtain a Biosciences), CD4 (clone GK1.5; BD Biosciences), CD8 (clone 53-6.7; eBioscience),
MERS-CoV that was adapted to cause respiratory disease in mice (MERS-15). At CD25 (clone PC61; BD Biosciences), CD26/DPP4 (clone H194-112; Biolegend),
each passage, the lungs were homogenized and 50 µl lung homogenate was used for CD69 (clone H1.2F3; eBioscience), interferon-γ (clone XMG1.2; BD Biosciences),
intranasal infection of a naïve 288/330+/− mouse. The MERS-15 mouse-adapted tumour-necrosis factor-α (clone MP6-XT22; eBioscience) and interleukin-2 (clone
virus was assessed in mice by survival, weight loss, lung titre, haemorrhaging, JES6-5H4; eBioscience). Cells were also treated with a fixable LIVE/DEAD
respiratory function and histopathology indicative of diffuse alveolar damage and discriminator (Invitrogen). Stained cells were fixed for 20 min in 1%
ARDS. Clonal isolates of the MERS-15 virus were obtained by plaque purification paraformaldehyde, resuspended in PBS and stored at 4 °C before acquisition within
and amplification in Vero CCL81 cells, and were sequenced to determine the 24 h. Cell populations were first gated to exclude (1) debris, (2) doublet events and
mutations acquired during mouse adaptation of the virus. All sequencing was (3) dead cells before phenotypic and functional gating. All samples were stained in
performed at the UNC-CH Genome Analysis Facility. MERS-15 C2 reproduced the parallel with controls of fluorescence – 1. A minimum of 100,000 live, singlet events
severe respiratory disease observed with MERS-15; therefore, all subsequent were acquired per sample, and data were analysed using FlowJo software (TreeStar).
experiments were performed with the plaque-purified MERS-15 C2.
Northern blot analysis and RT-PCR. Northern blot analysis was performed for
Human monoclonal antibody 3B11 protection study. The 288/330+/+ mice were detection of full-length mDPP4 in the lungs of 288/330+/+, 288/330+/− and C57BL/6J
prophylactically administered 250 µg of human monoclonal antibody 3B11 or wild-type mice. Poly(A) RNA was isolated to eliminate ribosomal RNA (rRNA;
isotype control antibody F10 by intraperitoneal injection19 12 h before infection with Qiagen). Equivalent amounts of RNA were resolved on 0.8% agarose gels and
MERS-15 C2. Mice were monitored daily for weight and survival, and killed at days 3 transferred to nitrocellulose membrane. A biotinylated probe (5′-biotin-gATg
and 6 p.i. to collect the lungs for histology, for viral titre assessment by plaque assay (biotin-dT)gCTggTgAgCTgTgCTgCTAgCgATCCCgTggTCTTCATCC-3′) was
and for evaluation of lung haemorrhaging. Respiratory function was measured at used to detect mDPP4.

NATURE MICROBIOLOGY 2, 16226 (2016) | DOI: 10.1038/nmicrobiol.2016.226 | www.nature.com/naturemicrobiology 9

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
ARTICLES NATURE MICROBIOLOGY

To quantify viral and targeted host mRNAs, MERS-CoV, mDPP4 and mouse References
glyceraldehyde 3-phosphate dehydrogenase (GAPDH) RNAs were measured in 1. Chan, J. F.-W. et al. Treatment with lopinavir/ritonavir or interferon-β1b
brain and lung tissue from MERS-CoV-infected mice. Briefly, tissues were removed improves outcome of MERS-CoV infection in a nonhuman primate model of
and placed into RNALater (Ambion) solution and stored at −80 °C until analysis by common marmoset. J. Infect. Dis. 212, 1904–1913 (2015).
RT-PCR. Lung tissue was homogenized in TRIzol reagent (Invitrogen) and isolated 2. de Wit, E. et al. Middle East respiratory syndrome coronavirus (MERS-CoV)
according to the manufacturer’s instructions. Standard RT-PCR was performed with causes transient lower respiratory tract infection in rhesus macaques. Proc. Natl
the following primer pairs for each of the indicated RNAs: MERS-CoV subgenomic Acad. Sci. USA 110, 16598–16603 (2013).
leader sequence: 5′-CTATCTCACTTCCCCTCgTTCTC-3′ and 5′- GAATCATTG 3. Falzarano, D. et al. Infection with MERS-CoV causes lethal pneumonia in the
TTAGGGTTCCG-3′; mouse GAPDH: 5′-CAACgACCCCTTCATTgACC-3′ and common marmoset. PLoS Pathogens 10, e1004250 (2014).
5′-GCAGGGATGATGTTCTGGGC-3′; and mDPP4: 5′-TAACGACACAGGAGTG 4. Munster, V. J., de Wit, E. & Feldmann, H. Pneumonia from human coronavirus
CCGC-3′ and 5′-TCTGCTTTTTGACTACAGGG-3′. Equivalent volumes of PCR in a macaque model. N. Engl. J. Med. 368, 1560–1562 (2013).
product were resolved on gels for a non-quantitative answer (presence or absence) of 5. Johnson, R. F. et al. 3B11-N, a monoclonal antibody against MERS-CoV, reduces
viral subgenomic transcripts in the brain and lung. lung pathology in rhesus monkeys following intratracheal inoculation of MERS-
Quantitative RT-PCR was carried out on a Roche LightCycler 480 II, with
CoV Jordan-n3/2012. Virology 490, 49–58 (2016).
accompanying software, to analyse MERS-CoV viral RNA and mDPP4 mRNAs, and
6. Johnson, R. F. et al. Intratracheal exposure of common marmosets to MERS-
18S rRNA as an endogenous control. All RNAs were reverse transcribed under
CoV Jordan-n3/2012 or MERS-CoV EMC/2012 isolates does not result in lethal
standard conditions in a 20 µl reaction volume using SuperScript III reverse
disease. Virology 485, 422–430 (2015).
transcriptase (Invitrogen). MERS-CoV viral RNA was assessed using the
7. Cockrell, A. S. et al. Mouse dipeptidyl peptidase 4 is not a functional receptor
following primers at 900 nM in a 20 µl reaction in a SYBR Green assay with 2×
for Middle East respiratory syndrome coronavirus infection. J. Virol. 88,
SsoAdvanced Universal SYBR Green Supermix (Biorad). The forward primer
5195–5199 (2014).
anneals at the MERS-CoV leader sequence in the 5′ untranslated region
(5′-GAATAGCTTGGCTATCTCAC-3′) and the reverse primer anneals in the N 8. Coleman, C. M., Matthews, K. L., Goicochea, L. & Frieman, M. B. Wild-type and
gene (5′-TTGTTATCGGCAAAGGAAAC-3′). PCR conditions were 45 cycles of innate immune-deficient mice are not susceptible to the Middle East respiratory
95 °C for 10 s, 59 °C for 10 s and 72 °C for 15 s. Standard Taqman conditions syndrome coronavirus. J. Gen. Virol. 95, 408–412 (2014).
were used to analyse expression of mDPP4 and 18S rRNAs. The following 9. de Wit, E. et al. The Middle East respiratory syndrome coronavirus (MERS-
primers were used for mDPP4: 5′-CCCCAAGACAGTGTGGATTC-3′ and CoV) does not replicate in Syrian hamsters. PLoS ONE 8, e69127 (2013).
5′-GAGGATGAGCTGAGAGAGTCTATATTT-3′; probe no. 51 was used from the 10. Raj, V. S. et al. Adenosine deaminase acts as a natural antagonist for dipeptidyl
Roche Universal ProbeLibrary. PCR conditions were 45 cycles of 95 °C for 15 s and peptidase 4-mediated entry of the Middle East respiratory syndrome
60 °C for 50 s. A 20× commercially available primer/probe set was utilized to coronavirus. J. Virol. 88, 1834–1838 (2014).
quantitate the 18S rRNA (Life Technologies). 11. Ohnuma, K., Dang, N. H. & Morimoto, C. Revisiting an old acquaintance:
CD26 and its molecular mechanisms in T cell function. Trends Immunol. 29,
Statistical analysis. All quantitative data are presented as means ± 1 s.d. Significance 295–301 (2008).
between specific data sets is described in the respective figure legends and was 12. Agrawal, A. S. et al. Generation of a transgenic mouse model of Middle East
determined by Student’s t-test using a one-tailed or two-tailed distribution in respiratory syndrome coronavirus infection and disease. J. Virol. 89,
Microsoft Excel software. 3659–3670 (2015).
13. Li, K. et al. Middle East respiratory syndrome coronavirus causes multiple organ
Biosafety and biosecurity. In vivo adaptation of the MERS-0 virus in mice had been damage and lethal disease in mice transgenic for human dipeptidyl peptidase 4.
executed before publication of the US Government’s statement on funding pause on J. Infect. Dis. 213, 712–722 (2016).
certain types of GOF research37. When notice to cease all in vivo passage 14. Zhao, G. et al. Multi-organ damage in human dipeptidyl peptidase 4 transgenic
experiments was received, all studies to adapt the MERS-0 virus in vivo were mice infected with Middle East respiratory syndrome-coronavirus. PLoS ONE
immediately halted. Following our formal written request for continuation, and on 10, e0145561 (2015).
receiving an exemption from the pause and approval to continue after a National 15. Zhao, J. et al. Rapid generation of a mouse model for Middle East respiratory
Institute of Allergy and Infectious Diseases/NIH review, the in vivo adaptation of syndrome. Proc. Natl Acad. Sci. USA 111, 4970–4975 (2014).
MERS-0 was then continued by serial passage in mice. All studies using MERS-CoV 16. Lambeir, A. M., Durinx, C., Scharpe, S. & de Meester, I. Dipeptidyl-peptidase IV
were executed in BSL3 facilities at UNC-CH, under conditions described previously35. from bench to bedside: an update on structural properties, functions, and clinical
The following biosafety and biosecurity paragraph is as described by aspects of the enzyme DPP IV. Crit. Rev. Clin. Lab. Sci. 40, 209–294 (2003).
Menachery et al. 35 All work for these studies was performed with approved standard 17. Menachery, V. D., Gralinski, L. E., Baric, R. S. & Ferris, M. T. New metrics for
operating procedures and safety conditions for MERS-CoV (not a select agent), evaluating viral respiratory pathogenesis. PLoS ONE 10, e0131451 (2015).
SARS-CoV (select agent) and derivatives therein. Our institutional CoV BSL3 18. Ng, D. L. et al. Clinicopathologic, immunohistochemical, and ultrastructural
facilities have been designed to conform to the safety requirements that are findings of a fatal case of Middle East respiratory syndrome coronavirus
recommended by the Biosafety in Microbiological and Biomedical Laboratories, the infection in the United Arab Emirates, April 2014. Am. J. Pathol. 186,
US Department of Health and Human Services, the Public Health Service, the 652–658 (2016).
Centers for Disease Control and the NIH. Laboratory safety plans were submitted to, 19. Tang, X. C. et al. Identification of human neutralizing antibodies against MERS-
and the facility has been approved for use by, the UNC Department of CoV and their role in virus adaptive evolution. Proc. Natl Acad. Sci. USA 111,
Environmental Health and Safety (EHS) and the CDC. Electronic card access is E2018–E2026 (2014).
required for entry into the facility. All workers have been trained by EHS to safely use 20. Agnihothram, S. et al. A mouse model for Betacoronavirus subgroup 2c using a
powered air-purifying respirators, and appropriate work habits in a BSL3 facility and bat coronavirus strain HKU5 variant. mBio 5, e00047-14 (2014).
active medical surveillance plans are in place. Our BSL3 facilities contain redundant 21. Korteweg, C. & Gu, J. Pathology, molecular biology, and pathogenesis of avian
fans, emergency power to fans, and biological safety cabinets and freezers, and our influenza A (H5N1) infection in humans. Am. J. Pathol. 172, 1155–1170 (2008).
facilities can accommodate SealSafe mouse racks. Materials classified as BSL3 agents 22. Ng, W.-F., To, K.-F., Lam, W. W., Ng, T.-K. & Lee, K.-C. The comparative
consist of MERS-CoV, SARS-CoV, bat CoV precursor strains and mutants derived pathology of severe acute respiratory syndrome and avian influenza A subtype
from these pathogens. Within the BSL3 facilities, experimentation with infectious H5N1 – a review. Hum. Pathol. 37, 381–390 (2006).
virus is performed in a certified Class II Biosafety Cabinet. All members of staff wear 23. Sanders, C. J. et al. Compromised respiratory function in lethal influenza
scrubs, Tyvek suits and aprons, powered air-purifying respirators and shoe covers, infection is characterized by the depletion of type I alveolar epithelial cells
and their hands are double-gloved. BSL3 users are subject to a medical surveillance beyond threshold levels. Am. J. Physiol. Lung Cell. Mol. Physiol. 304,
plan monitored by the University Employee Occupational Health Clinic (UEOHC), L481–L488 (2013).
which includes a yearly physical, annual influenza vaccination and mandatory 24. Pascal, K. E. et al. Pre- and postexposure efficacy of fully human antibodies
reporting of any symptoms associated with coronvirus infection during periods against Spike protein in a novel humanized mouse model of MERS-CoV
when working in the BSL3. All BSL3 users are trained in exposure management and
infection. Proc. Natl Acad. Sci. USA 112, 8738–8743 (2015).
reporting protocols, are prepared to self-quarantine and have been trained for safe
25. Frieman, M. et al. Molecular determinants of severe acute respiratory syndrome
delivery to a local infectious disease management department in an emergency
coronavirus pathogenesis and virulence in young and aged mouse models of
situation. All potential exposure events are reported and investigated by EHS and
human disease. J. Virol. 86, 884–897 (2012).
UEOHC, with reports filed to both the CDC and the NIH.
26. Thornbrough, J. M. et al. Middle East respiratory syndrome coronavirus NS4b
protein inhibits host RNase L activation. mBio 7, e00258 (2016).
Data availability. The data that support the findings of this study are available from
27. Dediego, M. L. et al. Pathogenicity of severe acute respiratory coronavirus
the corresponding author on request.
deletion mutants in hACE-2 transgenic mice. Virology 376, 379–389 (2008).
28. Sims, A. C. et al. Release of severe acute respiratory syndrome coronavirus
Received 27 April 2016; accepted 14 October 2016; nuclear import block enhances host transcription in human lung cells. J. Virol.
published 28 November 2016; corrected 14 July 2017 87, 3885–3902 (2013).

10 NATURE MICROBIOLOGY 2, 16226 (2016) | DOI: 10.1038/nmicrobiol.2016.226 | www.nature.com/naturemicrobiology

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
NATURE MICROBIOLOGY ARTICLES
29. Agrawal, A. S. et al. Passive transfer of a germline-like neutralizing human AI100625 (R.S.B. and M.T.H.). GOF research considerations involving MERS-0 in vivo
monoclonal antibody protects transgenic mice against lethal Middle East passage in mice and the current manuscript were both reviewed and approved by the
respiratory syndrome coronavirus infection. Sci. Rep. 6, 31629 (2016). funding agency, the NIH. The content is solely the responsibility of the authors and does
30. Laksitorini, M., Prasasty, V. D., Kiptoo, P. K. & Siahaan, T. J. Pathways and not necessarily represent the official views of the NIH. Generation of CRISPR–Cas9-
progress in improving drug delivery through the intestinal mucosa and blood– modified mice was performed at the UNC Animal Models Core Facility under the direction
brain barriers. Ther. Deliv. 5, 1143–1163 (2014). of Dale Cowley.
31. McCray, P. B. Jr et al. Lethal infection of K18-hACE2 mice infected with severe
acute respiratory syndrome coronavirus. J. Virol. 81, 813–821 (2007). Author contributions
32. Lamers, M. M. et al. Deletion variants of Middle East respiratory syndrome A.S.C. conceived/designed, coordinated and executed the experiments, analysed the data
coronavirus from humans, Jordan, 2015. Emerg. Infect. Dis. 22, 716–719 (2016). and wrote the manuscript. B.L.Y. developed and recovered infectious clone viruses. T.S.
33. Scobey, T. et al. Reverse genetics with a full-length infectious cDNA of the completed mouse experiments. K.J. designed and completed immunological experiments.
Middle East respiratory syndrome coronavirus. Proc. Natl Acad. Sci. USA 110, M.D. helped establish and maintain the mouse colony and perform molecular analyses.
16157–16162 (2013). A.B. helped complete the mouse experiments. X.-C.T. and W.A.M. provided critical
34. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, monoclonal antibody reagents. M.T.H. and R.S.B. conceived/designed the experiments and
823–826 (2013). wrote the manuscript.
35. Menachery, V. D. et al. A SARS-like cluster of circulating bat coronaviruses
shows potential for human emergence. Nat. Med. 21, 1508–1513 (2015).
36. Ayala, J. E. et al. Standard operating procedures for describing and performing Additional information
metabolic tests of glucose homeostasis in mice. Dis. Model. Mech. 3, Supplementary information is available for this paper.
525–534 (2010). Reprints and permissions information is available at www.nature.com/reprints.
37. US Government Deliberative Process Research Funding Pause on Selected Gain-of- Correspondence and requests for materials should be addressed to A.S.C., M.T.H. and R.S.B.
Function Research Involving Influenza, MERS, and SARS Viruses (US Government,
2014); http://www.phe.gov/s3/dualuse/Documents/gain-of-function.pdf How to cite this article: Cockrell, A. S. et al. A mouse model for MERS coronavirus-induced
acute respiratory distress syndrome. Nat. Microbiol. 2, 16226 (2016).
Acknowledgements
These studies were supported by grants from the National Institute of Allergy and Competing interests
Infectious Disease of the US NIH by awards HHSN272201000019I-HHSN27200003 W.A.M. has a financial interest in AbViro. The other authors declare no competing
(R.S.B. and M.T.H.), AI106772, AI108197, AI110700 and AI109761 (R.S.B.), and U19 financial interests.

NATURE MICROBIOLOGY 2, 16226 (2016) | DOI: 10.1038/nmicrobiol.2016.226 | www.nature.com/naturemicrobiology 11

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

You might also like